Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 593
Filter
1.
Nat Rev Nephrol ; 20(6): 371-385, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38443711

ABSTRACT

Podocytes form the backbone of the glomerular filtration barrier and are exposed to various mechanical forces throughout the lifetime of an individual. The highly dynamic biomechanical environment of the glomerular capillaries greatly influences the cell biology of podocytes and their pathophysiology. Throughout the past two decades, a holistic picture of podocyte cell biology has emerged, highlighting mechanobiological signalling pathways, cytoskeletal dynamics and cellular adhesion as key determinants of biomechanical resilience in podocytes. This biomechanical resilience is essential for the physiological function of podocytes, including the formation and maintenance of the glomerular filtration barrier. Podocytes integrate diverse biomechanical stimuli from their environment and adapt their biophysical properties accordingly. However, perturbations in biomechanical cues or the underlying podocyte mechanobiology can lead to glomerular dysfunction with severe clinical consequences, including proteinuria and glomerulosclerosis. As our mechanistic understanding of podocyte mechanobiology and its role in the pathogenesis of glomerular disease increases, new targets for podocyte-specific therapeutics will emerge. Treating glomerular diseases by targeting podocyte mechanobiology might improve therapeutic precision and efficacy, with potential to reduce the burden of chronic kidney disease on individuals and health-care systems alike.


Subject(s)
Podocytes , Podocytes/physiology , Humans , Biomechanical Phenomena , Mechanotransduction, Cellular/physiology , Cytoskeleton/physiology , Biophysics , Animals , Cell Adhesion/physiology
2.
Kidney Int ; 104(1): 33-35, 2023 07.
Article in English | MEDLINE | ID: mdl-37349059

ABSTRACT

The role of parietal epithelial cells (PECs) in kidney function and disease was recently revisited. Building on previous studies of human kidney tissue, in the current issue, Liu et al. further characterize PECs using single-cell RNA sequencing data and confirm the crucial pathophysiological role of PECs in murine kidney biology as a reservoir for different types of progenitors.


Subject(s)
Kidney Glomerulus , Podocytes , Humans , Mice , Animals , Podocytes/physiology , Epithelial Cells/physiology , Kidney
4.
Biochem Biophys Res Commun ; 614: 198-206, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35605301

ABSTRACT

Podocyte damage is a major pathological lesion leading to focal segmental glomerulosclerosis (FSGS). Podocytes damaged by cellular stress undergo hypertrophy to compensate for podocytopenia. It is known that cyclin-dependent kinase inhibitors induced by p53 ensure podocytes hypertrophy; however, its precise mechanism remains to be further investigated. In this study, we found that ubiquitin specific protease 40 (USP40) is a novel regulator of p53. Although USP40 knockout mice established in the present study revealed no abnormal kidney phenotype, intermediate filament Nestin was upregulated in the glomeruli, and was bound to and colocalized with USP40. We also found that USP40 deubiquitinated histidine triad nucleotide-binding protein 1 (HINT1), an inducer of p53. Gene knockdown experiments of USP40 in cultured podocytes revealed the reduction of HINT1 and p53 protein expression. Finally, in glomerular podocytes of mouse FSGS, upregulation of HINT1 occurred in advance of the proteinuria, which was followed by upregulation of USP40, p53 and Nestin. In conclusion, USP40 bound to Nestin deubiquitinates HINT1, and in consequence upregulates p53. These results provide additional insight into the pathological mechanism of podocyte hypertrophy in FSGS.


Subject(s)
Glomerulosclerosis, Focal Segmental , Nerve Tissue Proteins , Nestin , Podocytes , Tumor Suppressor Protein p53 , Ubiquitin-Specific Proteases , Animals , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/metabolism , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Hypertrophy , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Podocytes/metabolism , Podocytes/pathology , Podocytes/physiology , Protein Kinase C/antagonists & inhibitors , Stress, Physiological/genetics , Stress, Physiological/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Up-Regulation
5.
Clin Immunol ; 234: 108919, 2022 01.
Article in English | MEDLINE | ID: mdl-34974170

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease that affects almost any organ. Multiple immunological abnormalities involving every domain of the immune system contribute to the expression of the disease. It is now recognized that elements of the immune system instigate processes in tissue resident cells which execute organ damage. Although correction of ongoing immune aberrations is important in the control of disease activity, targeting tissue specific injurious processes may prove desirable in limiting organ damage.


Subject(s)
Lupus Erythematosus, Systemic/complications , Astrocytes/physiology , Humans , Keratinocytes/physiology , Langerhans Cells/physiology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/etiology , Microglia/physiology , Organ Specificity , Podocytes/physiology , Skin Diseases/etiology
6.
Int J Biochem Cell Biol ; 143: 106153, 2022 02.
Article in English | MEDLINE | ID: mdl-34974186

ABSTRACT

As part of the glomerular filtration membrane, podocyte is terminally differentiated, structurally unique, and highly specialized in maintaining kidney function. Proteinuria caused by podocyte injury (foot process effacement) is the clinical symptom of various kidney diseases (CKD), including nephrotic syndrome. Podocyte autophagy has become a powerful therapeutic strategy target in ameliorating podocyte injury. Autophagy is known to be associated significantly with sirtuin-1, proteinuria, and podocyte injury. Various key findings in podocyte autophagy were reported in the past ten years, such as the role of endoplasmic reticulum (ER) stress in podocyte autophagy impairment, podocyte autophagy-related gene, essential roles of the signaling pathways: Mammalian Target of Rapamycin (mTOR)/ Phosphoinositide 3-kinase (PI3k)/ serine/threonine kinase 1 (Akt) in podocyte autophagy. These significant factors caused podocyte injury associated with autophagy impairment. Sirtuin-1 was reported to have a vital key role in mTOR signaling, 5'AMP-activated protein kinase (AMPK) regulation, autophagy activation, and various critical pathways associated with podocyte's function and health; it has potential value to podocyte injury pathogenesis investigation. From these findings, podocyte autophagy has become an attractive therapeutic strategy to ameliorate podocyte injury, and this review will provide an in-depth review on therapeutic targets he podocyte autophagy.


Subject(s)
Autophagy/genetics , Podocytes/physiology , Signal Transduction/genetics , Animals , Disease Models, Animal , Humans , Male
7.
Tissue Eng Part B Rev ; 28(4): 938-948, 2022 08.
Article in English | MEDLINE | ID: mdl-34541902

ABSTRACT

Unraveling the complex behavior of healthy and disease podocytes by analyzing the changes in their unique arrangement of foot processes, slit diaphragm, and the three-dimensional (3D) morphology is a long-standing goal in kidney-glomerular research. The complexities surrounding the podocytes' accessibility in animal models and growing evidence of differences between humans and animal systems have compelled researchers to look for alternate approaches to study podocyte behaviors. With the advent of bioengineered models, an increasingly powerful and diverse set of tools is available to develop novel podocyte culture systems. This review discusses the pertinence of various culture models of podocytes to study podocyte mechanisms in both normal physiology and disease conditions. While no one in vitro system comprehensively recapitulates podocytes' in vivo architecture, we emphasize how the existing systems can be exploited to answer targeted questions on podocyte structure and function. We highlight the distinct advantages and limitations of using these models to study podocyte behaviors and screen therapeutics. Finally, we discuss various considerations and potential engineering strategies for developing next-generation complex 3D culture models for studying podocyte behaviors in vitro. Impact Statement In various glomerular kidney diseases, there are numerous alterations in podocyte structure and function. Yet, many of these disease events and the required targeted therapies remain unknown, resulting in nonspecific treatments. The scientific and clinical communities actively search for new modes to develop structurally and functionally relevant podocyte culture systems to gain insights into various diseases and develop therapeutics. Current in vitro systems help in some ways but are not sufficient. A deeper understanding of these previous approaches is essential to advance the field, and importantly, bioengineering strategies can contribute a unique toolbox to establish next-generation podocyte systems.


Subject(s)
Podocytes , Animals , Bioengineering , Humans , Kidney , Kidney Glomerulus , Podocytes/physiology
8.
Commun Biol ; 4(1): 1351, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857869

ABSTRACT

The glomerulus is the filtration unit of the kidney. Injury to any component of this specialised structure leads to impaired filtration and eventually fibrosis and chronic kidney disease. Current two and three dimensional (2D and 3D) models that attempt to recreate structure and interplay between glomerular cells are imperfect. Most 2D models are simplistic and unrepresentative, and 3D organoid approaches are currently difficult to reproduce at scale and do not fit well with current industrial drug-screening approaches. Here we report a rapidly generated and highly reproducible 3D co-culture spheroid model (GlomSpheres), better demonstrating the specialised physical and molecular structure of a glomerulus. Co-cultured using a magnetic spheroid formation approach, conditionally immortalised (CI) human podocytes and glomerular endothelial cells (GEnCs) deposited mature, organized isoforms of collagen IV and Laminin. We demonstrate a dramatic upregulation of key podocyte (podocin, nephrin and podocalyxin) and GEnC (pecam-1) markers. Electron microscopy revealed podocyte foot process interdigitation and endothelial vessel formation. Incubation with pro-fibrotic agents (TGF-ß1, Adriamycin) induced extracellular matrix (ECM) dysregulation and podocyte loss, which were attenuated by the anti-fibrotic agent Nintedanib. Incubation with plasma from patients with kidney disease induced acute podocyte loss and ECM dysregulation relative to patient matched remission plasma, and Nintedanib reduced podocyte loss. Finally, we developed a rapid imaging approach to demonstrate the model's usefulness in higher throughput pharmaceutical screening. GlomSpheres therefore represent a robust, scalable, replacement for 2D in vitro glomerular disease models.


Subject(s)
Coculture Techniques/methods , Drug Evaluation, Preclinical/methods , Kidney Glomerulus/physiology , Spheroids, Cellular/physiology , Cells, Cultured , Endothelial Cells/physiology , Humans , Podocytes/physiology
9.
Eur J Pharmacol ; 913: 174616, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34780752

ABSTRACT

High fructose has been reported to drive glomerular podocyte oxidative stress and then induce podocyte foot process effacement in vivo, which could be partly regarded as podocyte hypermotility in vitro. Atractylodin possesses anti-oxidative effect. The aim of this study was to explore whether atractylodin prevented against fructose-induced podocyte hypermotility via anti-oxidative property. In fructose-exposed conditionally immortalized human podocytes, we found that atractylodin inhibited podocyte hypermotility, and up-regulated slit diaphragm proteins podocin and nephrin, and cytoskeleton protein CD2-associated protein (CD2AP), α-Actinin-4 and synaptopodin expression, which were consistent with its anti-oxidative activity evidenced by up-regulation of catalase (CAT) and superoxide dismutase (SOD) 1 expression, and reduction of reactive oxygen species (ROS) production. Atractylodin also significantly suppressed expression of transient receptor potential channels 6 (TRPC6) and phosphorylated Ca2+/calmodulin-dependent protein kinase IV (CaMK4) in cultured podocytes with fructose exposure. Additionally, in fructose-exposed podocytes, CaMK4 siRNA up-regulated synaptopodin and reduced podocyte hypermotility, whereas, silencing of TRPC6 by siRNA decreased p-CaMK4 expression, inhibited podocyte hypermotility, showing TRPC6/p-CaMK4 signaling activation in podocyte hypermotility under fructose condition. Just like atractylodin, antioxidant N-acetyl-L-cysteine (NAC) could inhibit TRPC6/p-CaMK4 signaling activation to reduce fructose-induced podocytes hypermotility. These results first demonstrated that the anti-oxidative property of atractylodin may contribute to the suppression of podocyte hypermotility via inhibiting TRPC6/p-CaMK4 signaling and restoring synaptopodin expression abnormality.


Subject(s)
Antioxidants/pharmacology , Fructose/adverse effects , Furans/pharmacology , Podocytes/drug effects , Sweetening Agents/adverse effects , Calcium-Calmodulin-Dependent Protein Kinase Type 4/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cell Line , Cell Movement/drug effects , Down-Regulation/drug effects , Humans , Microfilament Proteins/metabolism , Oxidative Stress/drug effects , Phosphorylation , Podocytes/physiology , Proteolysis/drug effects , Signal Transduction/drug effects , TRPC6 Cation Channel/antagonists & inhibitors , TRPC6 Cation Channel/metabolism
10.
Mediators Inflamm ; 2021: 5220226, 2021.
Article in English | MEDLINE | ID: mdl-34616232

ABSTRACT

OBJECTIVE: Acute kidney injury (AKI) usually occurs during sepsis. Inflammation factors, such as high-mobility group box 1 (HMGB1), are dramatically upregulated under septic conditions. In our current work, the functions of HMGB1 in AKI were explored. METHODS: An AKI model was induced by the lipopolysaccharide (LPS) challenge in C57 mice. Podocytes were challenged by LPS for different durations. Subsequently, podocytes transfected with HMGB1 siRNA were exposed to LPS for 24 h. The expressions of supernatant HMGB1 and cellular active caspase-3 were examined by Western blotting analysis. To explore the effect of HMGB1 on tubular epithelial cells (TECs), HK-2 cells were exposed to HMGB1 at various concentrations for 24 h. Epithelial-mesenchymal transition (EMT) of HK-2 cells was evaluated by Western blotting analysis. Mitochondrial division and apoptosis of HK-2 cells were assessed by MitoTracker Red and Western blotting analysis, respectively. RESULTS: Compared with the sham control group, the expression of HMGB1 was increased in the kidney of AKI mice. Moreover, the expression of supernatant HMGB1 was increased in LPS-challenged podocytes compared with the control group. Knockdown of HMGB1 attenuated LPS-induced podocyte injury. Besides, EMT in TECs was triggered by HMGB1. Mitochondrial damage and apoptosis of HK-2 cells exposed to HMGB1 were markedly elevated compared with the control group. CONCLUSIONS: Collectively, HMGB1 release in podocytes was induced by LPS, subsequently leading to exacerbated AKI.


Subject(s)
Acute Kidney Injury/etiology , HMGB1 Protein/physiology , Lipopolysaccharides/pharmacology , Podocytes/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Epithelial-Mesenchymal Transition , Male , Mice , Mice, Inbred C57BL
11.
Int J Mol Sci ; 22(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299260

ABSTRACT

The CD73 pathway is an important anti-inflammatory mechanism in various disease settings. Observations in mouse models suggested that CD73 might have a protective role in kidney damage; however, no direct evidence of its role in human kidney disease has been described to date. Here, we hypothesized that podocyte injury in human kidney diseases alters CD73 expression that may facilitate the diagnosis of podocytopathies. We assessed the expression of CD73 and one of its functionally important targets, the C-C chemokine receptor type 2 (CCR2), in podocytes from kidney biopsies of 39 patients with podocytopathy (including focal segmental glomerulosclerosis (FSGS), minimal change disease (MCD), membranous glomerulonephritis (MGN) and amyloidosis) and a control group. Podocyte CD73 expression in each of the disease groups was significantly increased in comparison to controls (p < 0.001-p < 0.0001). Moreover, there was a marked negative correlation between CD73 and CCR2 expression, as confirmed by immunohistochemistry and immunofluorescence (Pearson r = -0.5068, p = 0.0031; Pearson r = -0.4705, p = 0.0313, respectively), thus suggesting a protective role of CD73 in kidney injury. Finally, we identify CD73 as a novel potential diagnostic marker of human podocytopathies, particularly of MCD that has been notorious for the lack of pathological features recognizable by light microscopy and immunohistochemistry.


Subject(s)
5'-Nucleotidase/genetics , Kidney Diseases/metabolism , Podocytes/metabolism , 5'-Nucleotidase/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases/physiopathology , Male , Middle Aged , Podocytes/physiology , Proteinuria , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
12.
J Am Soc Nephrol ; 32(8): 1974-1986, 2021 08.
Article in English | MEDLINE | ID: mdl-34078664

ABSTRACT

BACKGROUND: CKD is associated with the loss of functional nephr ons, leading to increased mechanical and metabolic stress in the remaining cells, particularly for cells constituting the filtration barrier, such as podocytes. The failure of podocytes to mount an adequate stress response can lead to further nephron loss and disease progression. However, the mechanisms that regulate this degenerative process in the kidney are unknown. METHODS: We combined in vitro, in vivo, and organ-on-chip approaches to identify the RE1-silencing transcription factor (REST), a repressor of neuronal genes during embryonic development, as a central regulator of podocyte adaptation to injury and aging. RESULTS: Mice with a specific deletion of REST in podocytes exhibit albuminuria, podocyte apoptosis, and glomerulosclerosis during aging, and exhibit increased vulnerability to renal injury. This phenotype is mediated, in part, by the effects of REST on the podocyte cytoskeleton that promote resistance to mechanical stressors and augment podocyte survival. Finally, REST expression is upregulated in human podocytes during aging, consistent with a conserved mechanism of stress resistance. CONCLUSIONS: These results suggest REST protects the kidney from injury and degeneration during aging, with potentially important therapeutic implications.


Subject(s)
Adaptation, Physiological/genetics , Aging/physiology , Podocytes/pathology , Podocytes/physiology , Repressor Proteins/genetics , Stress, Physiological/genetics , Adult , Aged , Aged, 80 and over , Albuminuria/genetics , Animals , Apoptosis/genetics , Cell Line , Cell Survival , Cytoskeleton/physiology , Gene Expression Regulation/genetics , Homeostasis/genetics , Humans , Mice , Phenotype , Repressor Proteins/metabolism , Sclerosis , Young Adult
13.
J Am Soc Nephrol ; 32(6): 1409-1424, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33795424

ABSTRACT

BACKGROUND: Podocyte slit diaphragms (SDs) are intercellular junctions that function as size-selective filters, excluding most proteins from urine. Abnormalities in SDs cause proteinuria and nephrotic syndrome. Podocytes exhibit apicobasal polarity, which can affect fundamental aspects of cell biology, including morphology, intercellular junction formation, and asymmetric protein distribution along the plasma membrane. Apical polarity protein mutations cause nephrotic syndrome, and data suggest apical polarity proteins regulate SD formation. However, there is no evidence that basolateral polarity proteins regulate SDs. Thus, the role of apicobasal polarity in podocytes remains unclear. METHODS: Genetic manipulations and transgenic reporters determined the effects of disrupting apicobasal polarity proteins in Drosophila nephrocytes, which have SDs similar to those of mammalian podocytes. Confocal and electron microscopy were used to characterize SD integrity after loss of basolateral polarity proteins, and genetic-interaction studies illuminated relationships among apicobasal polarity proteins. RESULTS: The study identified four novel regulators of nephrocyte SDs: Dlg, Lgl, Scrib, and Par-1. These proteins comprise the basolateral polarity module and its effector kinase. The data suggest these proteins work together, with apical polarity proteins, to regulate SDs by promoting normal endocytosis and trafficking of SD proteins. CONCLUSIONS: Given the recognized importance of apical polarity proteins and SD protein trafficking in podocytopathies, the findings connecting basolateral polarity proteins to these processes significantly advance our understanding of SD regulation.


Subject(s)
Cell Membrane/physiology , Cell Polarity , Drosophila/cytology , Intercellular Junctions , Membrane Proteins/metabolism , Podocytes/physiology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Endocytosis , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Intercellular Junctions/ultrastructure , Membrane Proteins/genetics , Microscopy, Confocal , Microscopy, Electron , Models, Biological , Podocytes/metabolism , Protein Transport , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vertebrates
14.
Int J Med Sci ; 18(10): 2093-2101, 2021.
Article in English | MEDLINE | ID: mdl-33859515

ABSTRACT

Aims: Long noncoding RNAs (lncRNAs) play key roles in the pathophysiology of DKD involving actions of microRNAs (miRNAs). The aims of the study were to establish the involvement of selected lncRNAs in the epigenetic mechanisms of podocyte damage and tubular injury in DKD of type 2 diabetes mellitus (DM) patients in relation to a particular miRNAs profile. Methods: A total of 136 patients with type 2 DM and 25 healthy subjects were assessed in a cross-sectional study concerning urinary albumin: creatinine ratio (UACR), eGFR, biomarkers of podocyte damage (synaptopodin, podocalyxin) and of proximal tubule (PT) dysfunction (Kidney injury molecule-1-KIM-1, N-acetyl-D-glucosaminidase-NAG), urinary lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), nuclear-enriched abundant transcript 1 (NEAT1), myocardial infarction-associated transcript (MIAT), taurine-upregulated gene 1 (TUG1), urinary miRNA21, 124, 93, 29a. Results: Multivariable regression analysis showed that urinary lncMALAT1 correlated directly with urinary synaptopodin, podocalyxin, KIM-1, NAG, miRNA21, 124, UACR, and negatively with eGFR, miRNA93, 29a (p<0.0001; R2=0.727); urinary lncNEAT1 correlated directly with synaptopodin, KIM-1, NAG, miRNA21, 124, and negatively with eGFR, miRNA93, 29a (p<0.0001; R2=0.702); urinary lncMIAT correlated directly with miRNA93 and 29a, eGFR (p<0.0001; R2=0.671) and negatively with synaptopodin, KIM-1, NAG, UACR, miRNA21, 124 (p<0.0001; R2=0.654); urinary lncTUG1 correlated directly with eGFR, miRNA93, 29a, and negatively with synaptopodin, podocalyxin, NAG, miRNA21, 124 (p<0.0001; R2=0.748). Conclusions: In patients with type 2 DM lncRNAs exert either deleterious or protective functions within glomeruli and PT. LncRNAs may contribute to DKD through modulating miRNAs expression and activities. This observation holds true independently of albuminuria and DKD stage.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/genetics , Kidney Tubules, Proximal/physiopathology , Podocytes/physiology , RNA, Long Noncoding/metabolism , Adult , Aged , Biomarkers/metabolism , Biomarkers/urine , Cross-Sectional Studies , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/urine , Diabetic Nephropathies/physiopathology , Diabetic Nephropathies/urine , Female , Gene Expression Regulation/physiology , Humans , Male , MicroRNAs/metabolism , Middle Aged , Protective Factors , RNA, Long Noncoding/urine , Risk Factors , Young Adult
15.
Life Sci ; 277: 119463, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33862110

ABSTRACT

Through the regulation of gene expression, microRNAs (miRNAs) are capable of modulating vital biological processes, such as proliferation, differentiation, and apoptosis. Several mechanisms control the function of miRNAs, including translational inhibition and targeted miRNA degradation. Through utilizing high-throughput screening methods, such as small RNA sequencing and microarray, alterations in miRNA expression of kidneys have recently been observed both in rodent models and humans throughout the development of chronic kidney disease (CKD) and acute kidney injury (AKI). The levels of miRNAs in urine supernatant, sediment, and exosomal fraction could predict novel biomarker candidates in different diseases of kidneys, including IgA nephropathy, lupus nephritis, and diabetic nephropathy. The therapeutic potential of administrating anti-miRNAs and miRNAs has also been reported recently. The present study is aimed at reviewing the state-of-the-art research with regards to miRNAs involved in renal disorders related to primary podocyte dysfunction by laying particular emphasis on Focal Segmental Glomerulosclerosis (FSGS), Minimal Change Disease (MCD) and Membranous Nephropathy (MN).


Subject(s)
Kidney Diseases/genetics , MicroRNAs/genetics , Podocytes/metabolism , Apoptosis/genetics , Biomarkers/metabolism , Diabetic Nephropathies/metabolism , Glomerulonephritis, Membranous/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Lupus Nephritis/metabolism , MicroRNAs/metabolism , Nephrosis, Lipoid/metabolism , Podocytes/physiology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism
17.
Nat Commun ; 12(1): 2277, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859189

ABSTRACT

Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene-regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Nephrons/growth & development , Organogenesis/genetics , Renal Insufficiency, Chronic/genetics , Animals , Cell Communication , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Epigenomics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genetic Loci/genetics , Genome-Wide Association Study , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Mice , Nephrons/cytology , Podocytes/physiology , Polymorphism, Single Nucleotide , RNA-Seq , Renal Insufficiency, Chronic/pathology , Single-Cell Analysis , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism
18.
J Am Soc Nephrol ; 32(6): 1389-1408, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33785583

ABSTRACT

BACKGROUND: Podocytes embrace the glomerular capillaries with foot processes, which are interconnected by a specialized adherens junction to ultimately form the filtration barrier. Altered adhesion and loss are common features of podocyte injury, which could be mediated by shedding of cell-adhesion molecules through the regulated activity of cell surface-expressed proteases. A Disintegrin and Metalloproteinase 10 (ADAM10) is such a protease known to mediate ectodomain shedding of adhesion molecules, among others. Here we evaluate the involvement of ADAM10 in the process of antibody-induced podocyte injury. METHODS: Membrane proteomics, immunoblotting, high-resolution microscopy, and immunogold electron microscopy were used to analyze human and murine podocyte ADAM10 expression in health and kidney injury. The functionality of ADAM10 ectodomain shedding for podocyte development and injury was analyzed, in vitro and in vivo, in the anti-podocyte nephritis (APN) model in podocyte-specific, ADAM10-deficient mice. RESULTS: ADAM10 is selectively localized at foot processes of murine podocytes and its expression is dispensable for podocyte development. Podocyte ADAM10 expression is induced in the setting of antibody-mediated injury in humans and mice. Podocyte ADAM10 deficiency attenuates the clinical course of APN and preserves the morphologic integrity of podocytes, despite subepithelial immune-deposit formation. Functionally, ADAM10-related ectodomain shedding results in cleavage of the cell-adhesion proteins N- and P-cadherin, thus decreasing their injury-related surface levels. This favors podocyte loss and the activation of downstream signaling events through the Wnt signaling pathway in an ADAM10-dependent manner. CONCLUSIONS: ADAM10-mediated ectodomain shedding of injury-related cadherins drives podocyte injury.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Nephritis/metabolism , Nephrotic Syndrome/metabolism , Podocytes/metabolism , Podocytes/pathology , Renal Insufficiency, Chronic/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Autoantibodies/adverse effects , Blood Urea Nitrogen , Cadherins/metabolism , Cell Adhesion , Cell Communication , Cell Membrane/metabolism , Cells, Cultured , Creatinine/urine , Disease Models, Animal , Female , Glomerular Filtration Barrier/pathology , Glomerular Filtration Barrier/physiopathology , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis/pathology , Nephrotic Syndrome/pathology , Podocytes/physiology , Proteomics , Tissue Array Analysis , Transcriptome , Wnt Signaling Pathway
19.
J Am Soc Nephrol ; 32(6): 1323-1337, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33771836

ABSTRACT

BACKGROUND: Transcriptional programs control cell fate, and identifying their components is critical for understanding diseases caused by cell lesion, such as podocytopathy. Although many transcription factors (TFs) are necessary for cell-state maintenance in glomeruli, their roles in transcriptional regulation are not well understood. METHODS: The distribution of H3K27ac histones in human glomerulus cells was analyzed to identify superenhancer-associated TFs, and ChIP-seq and transcriptomics were performed to elucidate the regulatory roles of the TFs. Transgenic animal models of disease were further investigated to confirm the roles of specific TFs in podocyte maintenance. RESULTS: Superenhancer distribution revealed a group of potential TFs in core regulatory circuits in human glomerulus cells, including FOXC1/2, WT1, and LMX1B. Integration of transcriptome and cistrome data of FOXC1/2 in mice resolved transcriptional regulation in podocyte maintenance. FOXC1/2 regulated differentiation-associated transcription in mature podocytes. In both humans and animal models, mature podocyte injury was accompanied by deregulation of FOXC1/2 expression, and FOXC1/2 overexpression could protect podocytes in zebrafish. CONCLUSIONS: FOXC1/2 maintain podocyte differentiation through transcriptional stabilization. The genome-wide chromatin resources support further investigation of TFs' regulatory roles in glomeruli transcription programs.


Subject(s)
Forkhead Transcription Factors/genetics , Podocytes/physiology , Transcription Factors/genetics , Transcription, Genetic , Animals , Cell Differentiation/genetics , Chromosome Mapping , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation , Histones , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Podocytes/pathology , Transcription Factors/metabolism , Transcriptome , WT1 Proteins/genetics , WT1 Proteins/metabolism , Zebrafish , Zebrafish Proteins/genetics
20.
J Hum Genet ; 66(8): 795-803, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33654185

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is a histological lesion with many causes, including inherited genetic defects, with significant proteinuria being the predominant clinical finding at presentation. FSGS is considered as a podocyte disease due to the fact that in the majority of patients with FSGS, the lesion results from defects in the podocyte structure. However, FSGS does not result exclusively from podocyte-associated genes. In this study, we used a genetic approach based on targeted next-generation sequencing (NGS) of 242 genes to identify the genetic cause of FSGS in seven Tunisian families. The sequencing results revealed the presence of eight distinct mutations including seven newly discovered ones: the c.538G>A (p.V180M) in NPHS2, c.5186G>A (p.R1729Q) in PLCE1 and c.232A>C (p.I78L) in PAX2 and five novel mutations in COL4A3 and COL4A4 genes. Four mutations (c.209G>A (p.G70D), c.725G>A (p.G242E), c.2225G>A (p.G742E), and c. 1681_1698del) were detected in COL4A3 gene and one mutation (c.1424G>A (p.G475D)) was found in COL4A4. In summary, NGS of a targeted gene panel is an ideal approach for the genetic testing of FSGS with multiple possible underlying etiologies. We have demonstrated that not only podocyte genes but also COL4A3/4 mutations should be considered in patients with FSGS.


Subject(s)
Autoantigens/genetics , Collagen Type IV/genetics , Collagen/genetics , Glomerulosclerosis, Focal Segmental/genetics , PAX2 Transcription Factor/genetics , Adult , Female , Genetic Diseases, Inborn/diagnosis , Genetic Predisposition to Disease , Genetic Testing/methods , Glomerulosclerosis, Focal Segmental/diagnosis , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Mutation, Missense , Pedigree , Podocytes/physiology , Tunisia , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...